Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlert

A major discovery of recent decades has been the existence of stem cells and their potential to repair many, if not most, tissues. With the aging population, many attempts have been made to use exogenous stem cells to promote tissue repair, so far with limited success. An alternative approach, which...

Full description

Bibliographic Details
Main Authors: Lee, G, Espirito Santo, A, Zwingenberger, S, Cai, L, Vogl, T, Feldmann, M, Horwood, N, Chan, J, Nanchahal, J
Format: Journal article
Published: National Academy of Sciences 2018
_version_ 1797074248752168960
author Lee, G
Espirito Santo, A
Zwingenberger, S
Cai, L
Vogl, T
Feldmann, M
Horwood, N
Chan, J
Nanchahal, J
author_facet Lee, G
Espirito Santo, A
Zwingenberger, S
Cai, L
Vogl, T
Feldmann, M
Horwood, N
Chan, J
Nanchahal, J
author_sort Lee, G
collection OXFORD
description A major discovery of recent decades has been the existence of stem cells and their potential to repair many, if not most, tissues. With the aging population, many attempts have been made to use exogenous stem cells to promote tissue repair, so far with limited success. An alternative approach, which may be more effective and far less costly, is to promote tissue regeneration by targeting endogenous stem cells. However, ways of enhancing endogenous stem cell function remain poorly defined. Injury leads to the release of danger signals which are known to modulate the immune response, but their role in stem cell-mediated repair in vivo remains to be clarified. Here we show that high mobility group box 1 (HMGB1) is released following fracture in both humans and mice, forms a heterocomplex with CXCL12, and acts via CXCR4 to accelerate skeletal, hematopoietic, and muscle regeneration in vivo. Pretreatment with HMGB1 2 wk before injury also accelerated tissue regeneration, indicating an acquired proregenerative signature. HMGB1 led to sustained increase in cell cycling in vivo, and using Hmgb1−/− mice we identified the underlying mechanism as the transition of multiple quiescent stem cells from G0 to GAlert. HMGB1 also transitions human stem and progenitor cells to GAlert. Therefore, exogenous HMGB1 may benefit patients in many clinical scenarios, including trauma, chemotherapy, and elective surgery.
first_indexed 2024-03-06T23:33:24Z
format Journal article
id oxford-uuid:6cd07b87-2994-4a83-801e-08243c46d6a6
institution University of Oxford
last_indexed 2024-03-06T23:33:24Z
publishDate 2018
publisher National Academy of Sciences
record_format dspace
spelling oxford-uuid:6cd07b87-2994-4a83-801e-08243c46d6a62022-03-26T19:13:40ZFully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlertJournal articlehttp://purl.org/coar/resource_type/c_dcae04bcuuid:6cd07b87-2994-4a83-801e-08243c46d6a6Symplectic Elements at OxfordNational Academy of Sciences2018Lee, GEspirito Santo, AZwingenberger, SCai, LVogl, TFeldmann, MHorwood, NChan, JNanchahal, JA major discovery of recent decades has been the existence of stem cells and their potential to repair many, if not most, tissues. With the aging population, many attempts have been made to use exogenous stem cells to promote tissue repair, so far with limited success. An alternative approach, which may be more effective and far less costly, is to promote tissue regeneration by targeting endogenous stem cells. However, ways of enhancing endogenous stem cell function remain poorly defined. Injury leads to the release of danger signals which are known to modulate the immune response, but their role in stem cell-mediated repair in vivo remains to be clarified. Here we show that high mobility group box 1 (HMGB1) is released following fracture in both humans and mice, forms a heterocomplex with CXCL12, and acts via CXCR4 to accelerate skeletal, hematopoietic, and muscle regeneration in vivo. Pretreatment with HMGB1 2 wk before injury also accelerated tissue regeneration, indicating an acquired proregenerative signature. HMGB1 led to sustained increase in cell cycling in vivo, and using Hmgb1−/− mice we identified the underlying mechanism as the transition of multiple quiescent stem cells from G0 to GAlert. HMGB1 also transitions human stem and progenitor cells to GAlert. Therefore, exogenous HMGB1 may benefit patients in many clinical scenarios, including trauma, chemotherapy, and elective surgery.
spellingShingle Lee, G
Espirito Santo, A
Zwingenberger, S
Cai, L
Vogl, T
Feldmann, M
Horwood, N
Chan, J
Nanchahal, J
Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlert
title Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlert
title_full Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlert
title_fullStr Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlert
title_full_unstemmed Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlert
title_short Fully reduced HMGB1 accelerates the regeneration of multiple tissues by transitioning stem cells to GAlert
title_sort fully reduced hmgb1 accelerates the regeneration of multiple tissues by transitioning stem cells to galert
work_keys_str_mv AT leeg fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert
AT espiritosantoa fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert
AT zwingenbergers fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert
AT cail fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert
AT voglt fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert
AT feldmannm fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert
AT horwoodn fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert
AT chanj fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert
AT nanchahalj fullyreducedhmgb1acceleratestheregenerationofmultipletissuesbytransitioningstemcellstogalert